Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
2.
Int J Mol Sci ; 22(3)2021 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-33513710

RESUMO

Stimulation of thermogenesis in brown adipose tissue (BAT) could have far-reaching health benefits in combatting obesity and obesity-related complications. Apolipoprotein A-IV (ApoA-IV), produced by the gut and the brain in the presence of dietary lipids, is a well-known short-term satiating protein. While our previous studies have demonstrated reduced diet-induced thermogenesis in ApoA-IV-deficient mice, it is unclear whether this reduction is due to a loss of peripheral or central effects of ApoA-IV. We hypothesized that central administration of ApoA-IV stimulates BAT thermogenesis and that sympathetic and sensory innervation is necessary for this action. To test this hypothesis, mice with unilateral denervation of interscapular BAT received central injections of recombinant ApoA-IV protein or artificial cerebrospinal fluid (CSF). The effects of central ApoA-IV on BAT temperature and thermogenesis in mice with unilateral denervation of the intrascapular BAT were monitored using transponder probe implantation, qPCR, and immunoblots. Relative to CSF, central administration of ApoA-IV significantly increased temperature and UCP expression in BAT. However, all of these effects were significantly attenuated or prevented in mice with unilateral denervation. Together, these results clearly demonstrate that ApoA-IV regulates BAT thermogenesis centrally, and this effect is mediated through sympathetic and sensory nerves.


Assuntos
Tecido Adiposo Marrom/fisiologia , Apolipoproteínas A/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema Nervoso Simpático/fisiologia , Termogênese/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Tecido Adiposo Marrom/enzimologia , Tecido Adiposo Marrom/metabolismo , Animais , Apolipoproteínas A/deficiência , Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Regulação da Expressão Gênica/genética , Lipase/genética , Lipase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes , Terceiro Ventrículo/fisiologia , Tirosina 3-Mono-Oxigenase/metabolismo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
3.
Biochem Biophys Res Commun ; 534: 659-665, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33239168

RESUMO

Apolipoprotein A4 (ApoA4) regulates lipid and glucose metabolism and exerts anti-inflammatory effects in atherogenesis and colitis. The present study explored the presumed protective role of ApoA4 in carbon tetrachloride (CCl4)-induced acute liver injury (ALI) in mice. The ALI model in wild type (WT), ApoA4 knock-out (ApoA4-KO) and ApoA4 transgenic (ApoA4-TG) mice was induced by a single intraperitoneal administration of CCl4. Liver and blood were harvested from mice to assess liver functions, immunohistological changes, immune cell populations and cytokine profiles. ApoA4 deficiency aggravated, and ApoA4 overexpression alleviated CCl4-inflicted liver damage by controlling levels of anti-oxidant enzymes. ApoA4 deletion increased the recruitment of monocytes/macrophages into the injured liver and upregulated the plasma levels of IL-6, TNF-α and MCP-1, but lower IL-10 and IFN-γ. ApoA4 over-expression rescued this effect and resulted in lower percentages of monocytes/macrophages and dendritic cells, the ratio of blood pro-inflammatory to anti-inflammatory monocytes and reduced plasma concentrations of IL-6, but enhanced IL-10 and IFN-γ. We propose ApoA4 as a potential new therapeutic target for the management of liver damage.


Assuntos
Apolipoproteínas A/metabolismo , Tetracloreto de Carbono/antagonistas & inibidores , Tetracloreto de Carbono/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Animais , Antioxidantes/metabolismo , Apolipoproteínas A/deficiência , Apolipoproteínas A/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocinas/sangue , Citocinas/genética , Mediadores da Inflamação/sangue , Fígado/efeitos dos fármacos , Fígado/imunologia , Fígado/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Camundongos Transgênicos , Monócitos/imunologia , Regulação para Cima
4.
Int J Mol Sci ; 20(13)2019 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-31261740

RESUMO

In the presence of dietary lipids, both apolipoprotein A-IV (ApoA-IV) production and brown adipose tissue (BAT) thermogenesis are increased. The effect of dietary lipid-induced AproA-IV on BAT thermogenesis and energy expenditure remains unknown. In the present study, we hypothesized that ApoA-IV knockout (ApoA-IV-KO) mice exhibited decreased BAT thermogenesis to affect energy homeostasis. To test this hypothesis, BAT thermogenesis in wildtype (WT) and ApoA-IV-KO mice fed either a standard low-fat chow diet or a high-fat diet (HFD) was investigated. When fed a chow diet, energy expenditure and food intake were comparable between WT and ApoA-IV-KO mice. After 1 week of HFD consumption, ApoA-IV-KO mice had comparable energy intake but produced lower energy expenditure relative to their WT controls in the dark phase. After an acute feeding of dietary lipids or 1-week HFD feeding, ApoA-IV-KO mice produced lower levels of uncoupling protein 1 (UCP1) and exhibited reduced expression of thermogenic genes in the BAT compared with WT controls. In response to cold exposure, however, ApoA-IV-KO mice had comparable energy expenditure and BAT temperature relative to WT mice. Thus, ApoA-IV-KO mice exhibited reduced diet-induced BAT thermogenesis and energy expenditure.


Assuntos
Tecido Adiposo Marrom/metabolismo , Apolipoproteínas A/genética , Dieta Hiperlipídica , Termogênese , Tecido Adiposo Marrom/fisiologia , Animais , Apolipoproteínas A/deficiência , Gorduras na Dieta/metabolismo , Metabolismo Energético , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Desacopladora 1/metabolismo
5.
Physiol Behav ; 188: 11-17, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29378187

RESUMO

Cholecystokinin (CCK) and apolipoprotein A-IV (ApoA-IV) are gastrointestinal peptides that play an important role in controlling energy homeostasis. Lymphatic ApoA-IV and plasma CCK secretion are mediated via a chylomicron formation-dependent pathway during a dietary lipid infusion. Given their similar roles as satiating proteins, the present study examines how the two peptides interact in their function. Specifically, this study sought to understand how ApoA-IV regulates CCK secretion. For this purpose, Cck gene expression in the small intestines of ApoA-IV knockout (ApoA-IV-KO) and wild-type (WT) mice were compared under an array of feeding conditions. When fed with a chow or high-fat diet (HFD), basal levels of Cck transcripts were significantly reduced in the duodenum of ApoA-IV-KO mice compared to WT mice. Furthermore, after an oral gavage of a lipid mixture, Cck gene expression in the duodenum was significantly reduced in ApoA-IV-KO mice relative to the change seen in WT mice. To determine the mechanism by which ApoA-IV modulates Cck gene expression, STC-1 cells were transfected with predesigned mouse lysophosphatidic acid receptor 5 (LPAR5) small interfering RNA (siRNA) to knockdown Lpar5 gene expression. In this in-vitro study, mouse recombinant ApoA-IV protein increased Cck gene expression in enteroendocrine STC-1 cells and stimulated CCK release from the STC-1 cells. However, the levels of CCK protein and Cck expression were attenuated when Lpar5 was knocked down in the STC-1 cells. Together these observations suggest that dietary lipid-induced ApoA-IV is associated with Cck synthesis in the duodenum and that ApoA-IV protein directly enhances CCK release through the activation of a LPAR5-dependent pathway.


Assuntos
Antioxidantes/farmacologia , Apolipoproteínas A/farmacologia , Colecistocinina/metabolismo , Duodeno/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Animais , Apolipoproteínas A/deficiência , Apolipoproteínas A/genética , Linhagem Celular Transformada , Colecistocinina/genética , Gorduras na Dieta/administração & dosagem , Relação Dose-Resposta a Droga , Duodeno/metabolismo , Regulação da Expressão Gênica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptores de Ácidos Lisofosfatídicos/metabolismo , Fatores de Tempo , Triglicerídeos/sangue
6.
Am J Physiol Regul Integr Comp Physiol ; 313(5): R535-R548, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28768657

RESUMO

Apolipoprotein AIV (ApoAIV) and cholecystokinin (CCK) are well-known satiating signals that are stimulated by fat consumption. Peripheral ApoAIV and CCK interact to prolong satiating signals. In the present study, we hypothesized that ApoAIV and CCK control energy homeostasis in response to high-fat diet feeding. To test this hypothesis, energy homeostasis in ApoAIV and CCK double knockout (ApoAIV/CCK-KO), ApoAIV knockout (ApoAIV-KO), and CCK knockout (CCK-KO) mice were monitored. When animals were maintained on a low-fat diet, ApoAIV/CCK-KO, ApoAIV-KO, and CCK-KO mice had comparable energy intake and expenditure, body weight, fat mass, fat absorption, and plasma parameters relative to the controls. In contrast, these KO mice exhibited impaired lipid transport to epididymal fat pads in response to intraduodenal infusion of dietary lipids. Furthermore, ApoAIV-KO mice had upregulated levels of CCK receptor 2 (CCK2R) in the small intestine while ApoAIV/CCK-KO mice had upregulated levels of CCK2R in the brown adipose tissue. After 20 wk of a high-fat diet, ApoAIV-KO and CCK-KO mice had comparable body weight and fat mass, as well as lower energy expenditure at some time points. However, ApoAIV/CCK-KO mice exhibited reduced body weight and adiposity relative to wild-type mice, despite having normal food intake. Furthermore, ApoAIV/CCK-KO mice displayed normal fat absorption and locomotor activity, as well as enhanced energy expenditure. These observations suggest that mice lacking ApoAIV and CCK have reduced body weight and adiposity, possibly due to impaired lipid transport and elevated energy expenditure.


Assuntos
Apolipoproteínas A/metabolismo , Colecistocinina/metabolismo , Gorduras na Dieta/metabolismo , Homeostase/fisiologia , Adiposidade/genética , Adiposidade/fisiologia , Animais , Apolipoproteínas A/deficiência , Peso Corporal/fisiologia , Colecistocinina/deficiência , Dieta com Restrição de Gorduras/métodos , Ingestão de Alimentos/fisiologia , Ingestão de Energia/fisiologia , Metabolismo Energético/genética , Camundongos Knockout
7.
J Lipid Res ; 57(3): 340-51, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26538546

RESUMO

Epidemiological, genetic association, and Mendelian randomization studies have provided strong evidence that lipoprotein (a) [Lp(a)] is an independent causal risk factor for CVD, including myocardial infarction, stroke, peripheral arterial disease, and calcific aortic valve stenosis. Lp(a) levels >50 mg/dl are highly prevalent (20% of the general population) and are overrepresented in patients with CVD and aortic stenosis. These data support the notion that Lp(a) should be a target of therapy for CVD event reduction and to reduce progression of aortic stenosis. However, effective therapies to specifically reduce plasma Lp(a) levels are lacking. Recent animal and human studies have shown that Lp(a) can be specifically targeted with second generation antisense oligonucleotides (ASOs) that inhibit apo(a) mRNA translation. In apo(a) transgenic mice, an apo(a) ASO reduced plasma apo(a)/Lp(a) levels and their associated oxidized phospholipid (OxPL) levels by 86 and 93%, respectively. In cynomolgus monkeys, a second generation apo(a) ASO, ISIS-APO(a)Rx, significantly reduced hepatic apo(a) mRNA expression and plasma Lp(a) levels by >80%. Finally, in a phase I study in normal volunteers, ISIS-APO(a)Rx ASO reduced Lp(a) levels and their associated OxPL levels up to 89 and 93%, respectively, with minimal effects on other lipoproteins. ISIS-APO(a)Rx represents the first specific and potent drug in clinical development to lower Lp(a) levels and may be beneficial in reducing CVD events and progression of calcific aortic valve stenosis.


Assuntos
Apolipoproteínas A/deficiência , Apolipoproteínas A/genética , Lipoproteína(a)/sangue , Oligonucleotídeos Antissenso/genética , Animais , Ensaios Clínicos como Assunto , Humanos
8.
J Lipid Res ; 52(11): 1984-94, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21840868

RESUMO

Apolipoprotein A-IV (apoA-IV) is synthesized by intestinal enterocytes during lipid absorption and secreted into lymph on the surface of nascent chylomicrons. A compelling body of evidence supports a central role of apoA-IV in facilitating intestinal lipid absorption and in regulating satiety, yet a longstanding conundrum is that no abnormalities in fat absorption, feeding behavior, or weight gain were observed in chow-fed apoA-IV knockout (A4KO) mice. Herein we reevaluated the impact of apoA-IV expression in C57BL6 and A4KO mice fed a high-fat diet. Fat balance and lymph cannulation studies found no effect of intestinal apoA-IV gene expression on the efficiency of fatty acid absorption, but gut sac transport studies revealed that apoA-IV differentially modulates lipid transport and the number and size of secreted triglyceride-rich lipoproteins in different anatomic regions of the small bowel. ApoA-IV gene deletion increased expression of other genes involved in chylomicron assembly, impaired the ability of A4KO mice to gain weight and increase adipose tissue mass, and increased the distal gut hormone response to a high-fat diet. Together these findings suggest that apoA-IV may play a unique role in integrating feeding behavior, intestinal lipid absorption, and energy storage.


Assuntos
Apolipoproteínas A/genética , Regulação da Expressão Gênica , Crescimento e Desenvolvimento/genética , Mucosa Intestinal/metabolismo , Metabolismo dos Lipídeos/genética , Tecido Adiposo/metabolismo , Animais , Apolipoproteínas A/química , Apolipoproteínas A/deficiência , Apolipoproteínas A/metabolismo , Transporte Biológico/genética , Peso Corporal/genética , Cateterismo , Dieta Hiperlipídica/efeitos adversos , Gorduras na Dieta/metabolismo , Ingestão de Alimentos/genética , Técnicas de Inativação de Genes , Peptídeo 1 Semelhante ao Glucagon/sangue , Vasos Linfáticos/cirurgia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tamanho da Partícula , Peptídeo YY/sangue , Triglicerídeos/metabolismo
9.
Inhal Toxicol ; 22(6): 449-59, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20235771

RESUMO

Environmental tobacco smoke (ETS) and ambient air fine particulate matter (PM(2.5)) are both complex mixtures that have important adverse effects on the cardiovascular system. Although exposures to these complex mixtures have been studied individually, direct comparisons between the two has not been performed. In this study, the authors employed a novel, noninvasive ultrasound biomicroscopy method (UBM) to assess the effects of long-term, low-concentration inhalations of side-stream smoke (SS) and concentrated ambient PM(2.5) (CAPs) on plaque progression. ApoE(-/-) mice (n = 8/group) on high-fat chow (HFC), or normal chow (NC), were exposed to SS (PM = 450 microg/m(3)) or filtered air (FA) for 6 h/day, 5 days/week, for 6 months; CAPs exposure was at 134 microg/m(3) (NC only). Mortality during the SS exposure was greater in the HFC than in the NC, and SS significantly enhanced the effects of diet. No mortality was observed in CAPs-exposed mice. At 4 and 6 months, SS produced the greatest change in plaque area in the left common carotid artery (CCA) in HFC as compared to FA or NC, but not in the brachiocephalic artery. In contrast, CAPs exposure significantly enhanced plaque areas in brachiocephalic and left CCA at 3 and 6 months of exposure. The effect of SS was comparable in magnitude to that produced by CAPs at an average PM(2.5) mass concentration that was only 30% as high. In light of the employment of the same animal model, uniform inhalation exposure protocols, time schedules, a noninvasive monitoring protocol, and a parallel study design, these findings have broad applicability.


Assuntos
Aterosclerose/etiologia , Exposição por Inalação/efeitos adversos , Material Particulado/toxicidade , Poluição por Fumaça de Tabaco/efeitos adversos , Envelhecimento/genética , Envelhecimento/patologia , Animais , Apolipoproteínas A/deficiência , Apolipoproteínas A/genética , Aterosclerose/diagnóstico por imagem , Aterosclerose/patologia , Dieta Aterogênica , Gorduras na Dieta/administração & dosagem , Modelos Animais de Doenças , Progressão da Doença , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tamanho da Partícula , Ultrassonografia
10.
J Intern Med ; 263(4): 450-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18324930

RESUMO

APOA5 encodes a novel apolipoprotein (apo A-V) which appears to be a modulator of plasma triglyceride (TG). In apoA5 knock out mice plasma TG level increases almost fourfold, whereas in human APOA5 transgenic mice it decreases by 70%. Some SNPs in the APOA5 gene have been associated with variations in plasma TG in humans. In addition, hypertriglyceridaemic (HTG) patients have been identified who carried rare nonsense mutations in the APOA5 gene (Q139X and Q148X), predicted to result in apo A-V deficiency. In this study we report a 17-year-old male with high TG and low high density lipoprotein cholesterol (HDL-C), who at the age of two had been found to have severe HTG and eruptive xanthomas suggesting a chylomicronaemia syndrome. Plasma postheparin LPL activity, however, was normal and no mutations were found in LPL and APOC2 genes. The sequence of APOA5 gene revealed that the patient was homozygous for a point mutation (c.289 C>T) in exon 4, converting glutamine codon at position 97 into a termination codon (Q97X). Apo A-V was not detected in patient's plasma, indicating that he had complete apo A-V deficiency. The administration of a low-fat and low-oligosaccharide diet, either alone or supplemented with omega-3 fatty acids, started early in life, reduced plasma TG to a great extent but had a negligible effect on plasma HDL-C. Loss of function mutations of APOA5 gene may be the cause of severe HTG in patients without mutations in LPL and APOC2 genes.


Assuntos
Apolipoproteínas A/deficiência , Apolipoproteínas A/genética , HDL-Colesterol/deficiência , Códon sem Sentido/genética , Hipertrigliceridemia/genética , Adolescente , Apolipoproteína A-V , Homozigoto , Humanos , Masculino
12.
Endocrinology ; 148(10): 4695-703, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17641001

RESUMO

Peptide YY (PYY)(3-36), released by intestinal lipid elicits functional effects that comprise the intestinal feedback response to luminal nutrients, but the pathway of action is not fully characterized. The aim of the present study was to determine the role of the apolipoprotein (apo) A-IV-cholecystokinin (CCK)(1) receptor (CCK(1)R) pathway in exogenous PYY(3-36)-induced activation of the gut-brain axis and inhibition of gastric emptying and food intake. PYY(3-36) (5 microg/100 g ip) significantly inhibited gastric emptying of a chow meal in wild-type but not A-IV(-/-) mice andCCK(1)R receptor blockade with devazepide (10 microg/100 g), abolished PYY(3-36)-induced inhibition of gastric emptying. PYY(3-36)-induced inhibition of food intake in both ad libitum-fed and 16-h fasted mice was unaltered in A-IV(-/-) mice, compared with wild-type controls, or by CCK(1)R receptor blockade with devazepide. PYY(3-36) activated neurons in the midregion of the nucleus of the solitary tract (bregma -7.32 to -7.76 mm) in A-IV(+/+) mice; this was measured by immunohistochemical localization of Fos protein. PYY(3-36)-induced Fos expression was significantly reduced by 65% in A-IV(+/+) mice pretreated systemically with the sensory neurotoxin capsaicin (5 mg/100 g), 78% by the CCK(1)R antagonist, devazepide (10 microg/100 g), and 39% by the Y2R antagonist, BIIE0246 (200 and 600 microg/100 g) and decreased by 67% in apo A-IV(-/-) mice, compared with A-IV(+/+) controls. The data suggest a role for apo A-IV and the CCK(1)R in PYY(3-36)-induced activation of the vagal afferent pathway and inhibition of gastric emptying, but this is likely not the pathway mediating the effects of PYY(3-36) on food intake.


Assuntos
Apolipoproteínas A/fisiologia , Retroalimentação Fisiológica/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Intestinos/fisiologia , Peptídeo YY/farmacologia , Receptor de Colecistocinina A/fisiologia , Ração Animal , Animais , Apolipoproteínas A/deficiência , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Jejum , Retroalimentação Fisiológica/fisiologia , Esvaziamento Gástrico/efeitos dos fármacos , Esvaziamento Gástrico/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Fragmentos de Peptídeos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Núcleo Solitário/citologia , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/metabolismo , Núcleo Solitário/fisiologia
14.
Curr Opin Lipidol ; 17(2): 122-7, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16531747

RESUMO

PURPOSE OF REVIEW: In this review we compare the phenotype and lipoprotein abnormalities of some patients who were found to carry mutations in the APOA5 gene predicted to result in apolipoprotein A-V deficiency. RECENT FINDINGS: The sequencing of the APOA5 gene in patients with primary hypertriglyceridemia, in whom mutations of the LPL and APOC2 genes had been excluded, led to the identification of four families with two different mutations in this gene predicted to result in truncated apolipoprotein A-V. The first mutation (Q148X) was found in a homozygous state in a child with severe type V hyperlipidemia, some clinical manifestations of chylomicronemia syndrome and a slight reduction in plasma postheparin lipoprotein lipase activity. Carriers of a different mutation (Q139X) were recently reported. Four Q139X heterozygotes had type V hyperlipidemia and markedly reduced plasma postheparin lipoprotein lipase activity. The hypertriglyceridemic Q139X heterozygote had other factors that could have contributed to hypertriglyceridemia. ApoB-100 kinetic studies in hypertriglyceridemic Q139X heterozygotes revealed an impairment of very low-density lipoprotein catabolism. SUMMARY: Mutations in the APOA5 gene, leading to truncated apolipoprotein A-V devoid of lipid-binding domains located in the carboxy-terminal end of the protein, if present in the homozygous state, are expected to cause severe type V hyperlipidemia in patients with no mutations in LPL or APOC2 genes. If present in the heterozygous state, these mutations predispose to hypertriglyceridemia in combination with other genetic factors or pathological conditions.


Assuntos
Apolipoproteínas A/deficiência , Apolipoproteínas A/genética , Triglicerídeos/metabolismo , Animais , Apolipoproteína A-V , Humanos , Lipase Lipoproteica/metabolismo
15.
J Lipid Res ; 47(5): 912-20, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16497661

RESUMO

Human data raised the possibility that coronary heart disease is associated with mutations in the apolipoprotein gene cluster APOA1/C3/A4 that result in multideficiency of cluster-encoded apolipoproteins and hypoalphalipoproteinemia. To test this hypothesis, we generated a mouse model for human apolipoprotein A-I (apoA-I)/C-III/A-IV deficiency. Homozygous mutants (Apoa1/c3/a4(-/-)) lacking the three cluster-encoded apolipoproteins were viable and fertile. In addition, feeding behavior and growth were apparently normal. Total cholesterol (TC), high density lipoprotein cholesterol (HDLc), and triglyceride levels in the plasma of fasted mutants fed a regular chow were 32% (P < 0.001), 17% (P < 0.001), and 70% (P < 0.01), respectively, those of wild-type mice. When fed a high-fat Western-type (HFW) diet, Apoa1/c3/a4(-/-) mice showed a further decrease in HDLc concentration and a moderate increase in TC, essentially in non-HDL fraction. The capacity of Apoa1/c3/a4(-/-) plasma to promote cholesterol efflux in vitro was decreased to 75% (P < 0.001), and LCAT activity was decreased by 38% (P < 0.01). Despite the very low total plasma cholesterol, the imbalance in lipoprotein distribution caused small but detectable aortic lesions in one-third of Apoa1/c3/a4(-/-) mice fed a HFW diet. In contrast, none of the wild-type mice had lesions. These results demonstrate that Apoa1/c3/a4(-/-) mice display clinical features similar to human apoA-I/C-III/A-IV deficiency (i.e., marked hypoalphalipoproteinemia) and provide further support for the apoa1/c3/a4 gene cluster as a minor susceptibility locus for atherosclerosis in mice.


Assuntos
Apolipoproteína A-I/deficiência , Apolipoproteínas A/deficiência , Apolipoproteínas C/deficiência , Gorduras na Dieta/efeitos adversos , Modelos Animais de Doenças , Dislipidemias/genética , Animais , Apolipoproteína C-III , Colesterol/metabolismo , Doença da Artéria Coronariana/patologia , Dislipidemias/patologia , Feminino , Humanos , Lipídeos/sangue , Lipoproteínas/sangue , Masculino , Camundongos , Ratos
16.
Atherosclerosis ; 164(2): 251-9, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12204795

RESUMO

Estrogen has previously been shown to inhibit development of early atherosclerotic lesions in hyperlipidemic mice. However, it is still not known whether estrogen also inhibits progression and destabilization of lesions once established and whether there are other effects of long-term hormone therapy in mice. To address this question, male, 20-week old, apolipoprotein E deficient mice were administered 17-beta estradiol or placebo subcutaneously for between 4 and 40 weeks. Estrogen administration did not cause regression of established lesions in the carotid arteries, aortic arch and thoracic aorta but prevented the initiation of new lesions in the abdominal aorta and iliac, femoral and popliteal arteries. Although the established lesions were slightly smaller in the innominate artery of the estrogen treated mice, estrogen did not prevent lesion progression. Estrogen administration also had no effect on the frequency of intra-plaque hemorrhage, atrophy of the fibrous cap, medial erosion, and fibro-fatty nodules, but did reduce the frequency of fatty streaks that form on top of or adjacent to the established lesions in the innominate artery. These data suggest that estrogen inhibits the initiation of the fatty streak but does not alter the progression of established lesions through stages of instability and healing.


Assuntos
Apolipoproteínas A/deficiência , Arteriosclerose/tratamento farmacológico , Arteriosclerose/patologia , Tronco Braquiocefálico/efeitos dos fármacos , Estradiol/farmacologia , Hemorragia/tratamento farmacológico , Hemorragia/patologia , Fosfatase Alcalina/sangue , Animais , Glicemia/análise , Tronco Braquiocefálico/patologia , Colesterol/sangue , Técnicas de Cultura , Modelos Animais de Doenças , Imuno-Histoquímica , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos , Probabilidade , Distribuição Aleatória , Valores de Referência , Sensibilidade e Especificidade , Estatísticas não Paramétricas
17.
J Assoc Physicians India ; 49: 274-8, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11225145

RESUMO

Deficiency of apolipoprotein can be of genetic origin or due to diseases like advanced chronic liver disease. Deficiency of apolipoprotein A causes Tangier disease without any major hepatic involvement being reported. Deficiency of apolipoprotein B causes abetalipoproteinemia or familial hypobetalipoproteinemia; with hepatic involvement in the form of raised transaminases, fatty liver and cirrhosis. Advanced chronic liver disease itself can cause reduction of apolipoprotein A and apolipoprotein B levels and acanthocytosis. In patients with chronic liver disease of undetermined etiology, lipid profile and apolipoprotein levels should be obtained routinely. If it suggests apolipoprotein B deficiency, then liver biopsy can be avoided, as the etiology of chronic liver disease is established. Isolated deficiency of either apolipoprotein A or apolipoprotein B suggests etiology of chronic liver disease, while deficiency of both apolipoprotein A and apolipoprotein B is a manifestation of advanced chronic liver disease.


Assuntos
Apolipoproteínas A/deficiência , Apolipoproteínas B/deficiência , Hepatopatias/etiologia , Colestase/patologia , Humanos , Fígado/patologia , Hepatopatias/patologia , Índice de Gravidade de Doença
18.
J Lipid Res ; 38(9): 1782-94, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9323588

RESUMO

To determine the physiological role of apolipoprotein (apo) A-IV, knockout mice were created by gene targeting in embryonic stem cells. In apoA-IV knockout mice, plasma cholesterol and triglyceride levels were reduced 25% and 44%, respectively, compared with controls. These changes were accounted for by decreased high density (HDL) and very low density lipoprotein (VLDL) levels, respectively, and metabolic studies indicated increased HDL-cholesteryl ester (CE) fractional catabolic rate (FCR) and reduced VLDL transport rate (TR), respectively. ApoA-IV knockout mice had greater than 70% reductions in both hepatic and intestinal apoC-III RNA levels and a similar reduction in the plasma apoC-III level. Complementation analysis, via crossbreeding of a mouse apoC-III transgene onto both the normal and apoA-IV knockout backgrounds, clearly demonstrated that the low triglyceride (VLDL) level in the apoA-IV knockout mice was due to alterations in apoC-III and not apoA-IV. ApoA-IV knockout mice had normal growth, feeding behavior, and lipid absorption, except male mice showed increased food intake in the 2 h after an 18-h fast, suggesting that under some circumstances apoA-IV might serve as a satiety factor. In summary, studies in apoA-IV-induced mutant mice have demonstrated a role for apoA-IV in increasing HDL cholesterol by inhibiting HDL cholesteryl ester FCR yet argue against the apolipoprotein as an overall important mediator of lipid absorption/metabolism.


Assuntos
Apolipoproteínas A/deficiência , HDL-Colesterol/sangue , Metabolismo dos Lipídeos , Animais , Apolipoproteína A-I/sangue , Apolipoproteína A-I/genética , Apolipoproteína C-III , Apolipoproteínas A/genética , Apolipoproteínas C/genética , Ésteres do Colesterol/sangue , HDL-Colesterol/metabolismo , Gorduras na Dieta/farmacocinética , Ingestão de Alimentos , Comportamento Alimentar , Feminino , Expressão Gênica , Crescimento , Absorção Intestinal , Lipídeos/sangue , Lipoproteínas/sangue , Lipoproteínas HDL/sangue , Lipoproteínas VLDL/sangue , Lipoproteínas VLDL/metabolismo , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , Aumento de Peso
19.
Nihon Rinsho ; 52(12): 3253-6, 1994 Dec.
Artigo em Japonês | MEDLINE | ID: mdl-7853719

RESUMO

The genes coding for apolipoprotein A-I, apo C-III, A-IV are located on the long arm of chromosome 11. The three genes are tandemly organized. Familial apolipoprotein A-I, C-III, A-IV deficiency was reported. The homozygous proband had extremely low HDL-cholesterol and premature coronary atherosclerosis. Analysis of the abnormal allele revealed the complete deletion of the apo A-I, C-III, A-IV genes. The deletion occurred in areas of repetitive DNA sequence. Familial apolipoprotein A-I, C-III deficiency has been shown to be due to the DNA inversion between the exon 4 of apo A-I and the first intron of apo C-III. The homozygous proband has HDL, deficiency and severe atherosclerosis. RFLP in this gene lesion has been reported and is closely related with the decrease of HDL, hypertriglyceridemia and coronary atherosclerosis. The elucidation of the relation between one particular allele in the lesion of apo A-I, C-III, A-IV gene cluster with lipoprotein metabolism and coronary atherosclerosis will require further determination of the precise nucleotide sequence variation.


Assuntos
Apolipoproteína A-I/deficiência , Apolipoproteínas A/deficiência , Apolipoproteínas C/deficiência , Apolipoproteína A-I/genética , Apolipoproteína C-III , Apolipoproteínas A/genética , Apolipoproteínas C/genética , Sequência de Bases , HDL-Colesterol/deficiência , Doença da Artéria Coronariana/etiologia , Deleção de Genes , Humanos , Dados de Sequência Molecular , Sequências Repetitivas de Ácido Nucleico
20.
Proc Natl Acad Sci U S A ; 88(7): 2793-7, 1991 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-1901417

RESUMO

The molecular genetic defect of a female patient with apolipoprotein A-I (apoA-I) deficiency and premature atherosclerosis was examined. Her parents were first cousins. Her plasma density fraction from 1.063 to 1.21 g/ml contained no apoA-I on SDS/PAGE and no measurable high density lipoprotein cholesterol. Southern blot hybridization showed no gross abnormality to be present in the patient's apoA-I gene and homozygosity for a haplotype of restriction fragment length polymorphisms in the apoA-I gene region. Sequencing after amplification by PCR revealed a codon 84 nonsense mutation (CAG----TAG, Gln----stop) of exon 4 and a codon 67 missense mutation (GCC----ACC, Ala----Thr) of exon 3 in the patient's apoA-I gene. The data from dot-blot hybridization with allele-specific oligonucleotide probes indicated that she was homozygous for the apoA-I gene with regard to the two mutations. The codon 37 missense mutation was also detected in the apoA-I gene of 6 out of 60 controls, who all had normal levels of apoA-I and high density lipoprotein cholesterol, suggesting that the missense mutation is polymorphic and not associated with apoA-I deficiency. These findings indicate that homozygosity for the apoA-I gene with codon 84 nonsense mutation causes the deficiency of apoA-I and of high density lipoprotein cholesterol in the patient.


Assuntos
Apolipoproteínas A/genética , Arteriosclerose/genética , Genes , Lipoproteínas HDL/genética , Mutação , Adulto , Sequência de Aminoácidos , Apolipoproteína A-I , Apolipoproteínas A/sangue , Apolipoproteínas A/deficiência , Arteriosclerose/diagnóstico por imagem , Sequência de Bases , Southern Blotting , Clonagem Molecular , Códon/genética , Angiografia Coronária , DNA/genética , DNA/isolamento & purificação , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Hibridização de Ácido Nucleico , Sondas de Oligonucleotídeos , Linhagem , Reação em Cadeia da Polimerase , Valores de Referência , Mapeamento por Restrição , Homologia de Sequência do Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA